Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Hepatol ; 72(4): 746-760, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31760071

RESUMEN

BACKGROUND & AIMS: Since human induced pluripotent stem cells (iPSCs) develop into hepatic organoids through stages that resemble human embryonic liver development, they can be used to study developmental processes and disease pathology. Therefore, we examined the early stages of hepatic organoid formation to identify key pathways affecting early liver development. METHODS: Single-cell RNA-sequencing and metabolomic analysis was performed on developing organoid cultures at the iPSC, hepatoblast (day 9) and mature organoid stage. The importance of the phosphatidylethanolamine biosynthesis pathway to early liver development was examined in developing organoid cultures using iPSC with a CRISPR-mediated gene knockout and an over the counter medication (meclizine) that inhibits the rate-limiting enzyme in this pathway. Meclizine's effect on the growth of a human hepatocarcinoma cell line in a xenotransplantation model and on the growth of acute myeloid leukemia cells in vitro was also examined. RESULTS: Transcriptomic and metabolomic analysis of organoid development indicated that the phosphatidylethanolamine biosynthesis pathway is essential for early liver development. Unexpectedly, early hepatoblasts were selectively sensitive to the cytotoxic effect of meclizine. We demonstrate that meclizine could be repurposed for use in a new synergistic combination therapy for primary liver cancer: a glycolysis inhibitor reprograms cancer cell metabolism to make it susceptible to the cytotoxic effect of meclizine. This combination inhibited the growth of a human liver carcinoma cell line in vitro and in a xenotransplantation model, without causing significant side effects. This drug combination was also highly active against acute myeloid leukemia cells. CONCLUSION: Our data indicate that phosphatidylethanolamine biosynthesis is a targetable pathway for cancer; meclizine may have clinical efficacy as a repurposed anti-cancer drug when used as part of a new combination therapy. LAY SUMMARY: The early stages of human liver development were modeled using human hepatic organoids. We identified a pathway that was essential for early liver development. Based upon this finding, a novel combination drug therapy was identified that could be used to treat primary liver cancer and possibly other types of cancer.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Leucemia Mieloide Aguda/sangre , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Meclizina/administración & dosificación , Fosfatidiletanolaminas/antagonistas & inhibidores , Fosfatidiletanolaminas/biosíntesis , Piridinas/administración & dosificación , Quinolinas/administración & dosificación , Adulto , Anciano , Animales , Carcinoma Hepatocelular/patología , Supervivencia Celular/efectos de los fármacos , Quimioterapia Combinada/métodos , Femenino , Técnicas de Inactivación de Genes , Glucólisis/efectos de los fármacos , Células Hep G2 , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Hígado/embriología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Organogénesis/efectos de los fármacos , Organogénesis/genética , Organoides/efectos de los fármacos , Organoides/metabolismo , ARN Nucleotidiltransferasas/deficiencia , ARN Nucleotidiltransferasas/genética , Estudios Retrospectivos , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L703-L709, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28258106

RESUMEN

There is considerable biological and physiological heterogeneity among patients who meet standard clinical criteria for acute respiratory distress syndrome (ARDS). In this study, we tested the hypothesis that there exists a subgroup of ARDS patients who exhibit a metabolically distinct profile. We examined undiluted pulmonary edema fluid obtained at the time of endotracheal intubation from 16 clinically phenotyped ARDS patients and 13 control patients with hydrostatic pulmonary edema. Nontargeted metabolic profiling was carried out on the undiluted edema fluid. Univariate and multivariate statistical analyses including principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA) were conducted to find discriminant metabolites. Seven-hundred and sixty unique metabolites were identified in the pulmonary edema fluid of these 29 patients. We found that a subset of ARDS patients (6/16, 38%) presented a distinct metabolic profile with the overrepresentation of 235 metabolites compared with edema fluid from the other 10 ARDS patients, whose edema fluid metabolic profile was indistinguishable from those of the 13 control patients with hydrostatic edema. This "high metabolite" endotype was characterized by higher concentrations of metabolites belonging to all of the main metabolic classes including lipids, amino acids, and carbohydrates. This distinct group with high metabolite levels in the edema fluid was also associated with a higher mortality rate. Thus metabolic profiling of the edema fluid of ARDS patients supports the hypothesis that there is considerable biological heterogeneity among ARDS patients who meet standard clinical and physiological criteria for ARDS.


Asunto(s)
Líquidos Corporales/metabolismo , Metabolómica/métodos , Edema Pulmonar/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Adulto , Análisis por Conglomerados , Demografía , Análisis Discriminante , Femenino , Humanos , Análisis de los Mínimos Cuadrados , Masculino , Metaboloma , Persona de Mediana Edad , Análisis de Componente Principal , Regulación hacia Arriba
3.
PLoS Med ; 12(2): e1001782, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25647612

RESUMEN

BACKGROUND: We know very little about the genetic factors affecting susceptibility to drug-induced central nervous system (CNS) toxicities, and this has limited our ability to optimally utilize existing drugs or to develop new drugs for CNS disorders. For example, haloperidol is a potent dopamine antagonist that is used to treat psychotic disorders, but 50% of treated patients develop characteristic extrapyramidal symptoms caused by haloperidol-induced toxicity (HIT), which limits its clinical utility. We do not have any information about the genetic factors affecting this drug-induced toxicity. HIT in humans is directly mirrored in a murine genetic model, where inbred mouse strains are differentially susceptible to HIT. Therefore, we genetically analyzed this murine model and performed a translational human genetic association study. METHODS AND FINDINGS: A whole genome SNP database and computational genetic mapping were used to analyze the murine genetic model of HIT. Guided by the mouse genetic analysis, we demonstrate that genetic variation within an ABC-drug efflux transporter (Abcb5) affected susceptibility to HIT. In situ hybridization results reveal that Abcb5 is expressed in brain capillaries, and by cerebellar Purkinje cells. We also analyzed chromosome substitution strains, imaged haloperidol abundance in brain tissue sections and directly measured haloperidol (and its metabolite) levels in brain, and characterized Abcb5 knockout mice. Our results demonstrate that Abcb5 is part of the blood-brain barrier; it affects susceptibility to HIT by altering the brain concentration of haloperidol. Moreover, a genetic association study in a haloperidol-treated human cohort indicates that human ABCB5 alleles had a time-dependent effect on susceptibility to individual and combined measures of HIT. Abcb5 alleles are pharmacogenetic factors that affect susceptibility to HIT, but it is likely that additional pharmacogenetic susceptibility factors will be discovered. CONCLUSIONS: ABCB5 alleles alter susceptibility to HIT in mouse and humans. This discovery leads to a new model that (at least in part) explains inter-individual differences in susceptibility to a drug-induced CNS toxicity.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/genética , Alelos , Encéfalo/metabolismo , Haloperidol/toxicidad , Síndromes de Neurotoxicidad/genética , Polimorfismo de Nucleótido Simple , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Adolescente , Adulto , Animales , Antipsicóticos/toxicidad , Barrera Hematoencefálica/metabolismo , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Ratones , Persona de Mediana Edad , Adulto Joven
4.
J Pharmacol Exp Ther ; 354(1): 73-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25962391

RESUMEN

Interspecies differences have limited the predictive utility of toxicology studies performed using animal species. A drug that could be a safe and effective treatment in humans could cause toxicity in animals, preventing it from being used in humans. We investigated whether the use of thymidine kinase (TK)-NOG mice with humanized livers could prevent this unfortunate outcome (i.e., "rescue" a drug for use in humans). A high dose of furosemide is known to cause severe liver toxicity in mice, but it is a safe and effective treatment in humans. We demonstrate that administration of a high dose of furosemide (200 mg/kg i.p.) causes extensive hepatotoxicity in control mice but not in humanized TK-NOG mice. This interspecies difference results from a higher rate of production of the toxicity-causing metabolite by mouse liver. Comparison of their survival curves indicated that the humanized mice were more resistant than control mice to the hepatotoxicity caused by high doses of furosemide. In this test case, humanized TK-NOG mouse studies indicate that humans could be safely treated with a high dose of furosemide.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Furosemida/toxicidad , Hepatocitos/patología , Hígado/efectos de los fármacos , Timidina Quinasa/genética , Animales , Relación Dosis-Respuesta a Droga , Furosemida/administración & dosificación , Furosemida/farmacocinética , Hepatocitos/trasplante , Humanos , Hígado/patología , Masculino , Ratones , Necrosis , Especificidad de la Especie , Distribución Tisular
5.
J Pharmacol Exp Ther ; 352(2): 274-80, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25424997

RESUMEN

Due to the substantial interspecies differences in drug metabolism and disposition, drug-induced liver injury (DILI) in humans is often not predicted by studies performed in animal species. For example, a drug (bosentan) used to treat pulmonary artery hypertension caused unexpected cholestatic liver toxicity in humans, which was not predicted by preclinical toxicology studies in multiple animal species. In this study, we demonstrate that NOG mice expressing a thymidine kinase transgene (TK-NOG) with humanized livers have a humanized profile of biliary excretion of a test (cefmetazole) drug, which was shown by an in situ perfusion study to result from interspecies differences in the rate of biliary transport and in liver retention of this drug. We also found that readily detectable cholestatic liver injury develops in TK-NOG mice with humanized livers after 1 week of treatment with bosentan (160, 32, or 6 mg/kg per day by mouth), whereas liver toxicity did not develop in control mice after 1 month of treatment. The laboratory and histologic features of bosentan-induced liver toxicity in humanized mice mirrored that of human subjects. Because DILI has become a significant public health problem, drug safety could be improved if preclinical toxicology studies were performed using humanized TK-NOG.


Asunto(s)
Cefmetazol/farmacocinética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Colestasis/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , Timidina Quinasa/genética , Animales , Bosentán , Enfermedad Hepática Inducida por Sustancias y Drogas/complicaciones , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Colestasis/etiología , Colestasis/patología , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ganciclovir/administración & dosificación , Ganciclovir/farmacología , Hepatocitos/metabolismo , Hepatocitos/fisiología , Hepatocitos/trasplante , Humanos , Tasa de Depuración Metabólica , Especificidad de la Especie , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Sulfonamidas/toxicidad , Timidina Quinasa/metabolismo , Distribución Tisular , Transgenes
6.
Pharmacogenet Genomics ; 23(2): 78-83, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23241944

RESUMEN

OBJECTIVE: We investigated whether human pharmacogenetic factors could be characterized using chimeric NOG mice expressing a thymidine kinase transgene (TK-NOG) with 'humanized' livers. MATERIALS AND METHODS: The rate of human-specific metabolism of two drugs was measured in chimeric mice reconstituted with human hepatocytes with different CYP2C19 and CYP2C9 genotypes. RESULTS: The rate of generation of human-predominant drug metabolites for S-mephenytoin and diclofenac in the chimeric mice was correlated with the CYP2C19 (n=9 donors, P=0.0005) or CYP2C9 (n=7 donors, P=0.0394) genotype, respectively, of the transplanted human hepatocytes. CONCLUSION: This study suggests that TK-NOG mice reconstituted with hepatocytes obtained from a relatively small number (3-10 per genotype) of human donors may be a promising model to identify human pharmacogenetic factors affecting the metabolism of clinically important drugs. For certain compounds, this innovative model system enables pharmacogenetic analyses to be efficiently performed in vivo within a human context and with control of all confounding environmental variables.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas/genética , Diclofenaco/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Mefenitoína/metabolismo , Farmacogenética , Animales , Proteínas Portadoras/fisiología , Células Cultivadas , Quimera/genética , Quimera/metabolismo , Citocromo P-450 CYP2C19 , Citocromo P-450 CYP2C9 , Hepatocitos/trasplante , Humanos , Hígado/citología , Ratones , Ratones Transgénicos , Timidina Quinasa/fisiología
7.
J Pharmacol Exp Ther ; 344(2): 388-96, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23143674

RESUMEN

Interspecies differences in drug metabolism have made it difficult to use preclinical animal testing data to predict the drug metabolites or potential drug-drug interactions (DDIs) that will occur in humans. Although chimeric mice with humanized livers can produce known human metabolites for test substrates, we do not know whether chimeric mice can be used to prospectively predict human drug metabolism or a possible DDI. Therefore, we investigated whether they could provide a more predictive assessment for clemizole, a drug in clinical development for the treatment of hepatitis C virus (HCV) infection. Our results demonstrate, for the first time, that analyses performed in chimeric mice can correctly identify the predominant human drug metabolite before human testing. The differences in the rodent and human pathways for clemizole metabolism were of importance, because the predominant human metabolite was found to have synergistic anti-HCV activity. Moreover, studies in chimeric mice also correctly predicted that a DDI would occur in humans when clemizole was coadministered with a CYP3A4 inhibitor. These results demonstrate that using chimeric mice can improve the quality of preclinical drug assessment.


Asunto(s)
Antivirales/metabolismo , Bencimidazoles/metabolismo , Hígado , Quimera por Trasplante/metabolismo , Animales , Antivirales/sangre , Antivirales/farmacocinética , Antivirales/uso terapéutico , Bencimidazoles/sangre , Bencimidazoles/farmacocinética , Bencimidazoles/uso terapéutico , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Semivida , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hepatitis C/enzimología , Hepatitis C/virología , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Valor Predictivo de las Pruebas , Ratas , Ritonavir/metabolismo , Ritonavir/farmacocinética , Ritonavir/farmacología , Especificidad de la Especie , Replicación Viral/efectos de los fármacos
8.
Gastrointest Endosc ; 78(2): 295-302.e2, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23566642

RESUMEN

BACKGROUND: Better pancreatic cyst fluid biomarkers are needed. OBJECTIVE: To determine whether metabolomic profiling of pancreatic cyst fluid would yield clinically useful cyst fluid biomarkers. DESIGN: Retrospective study. SETTING: Tertiary-care referral center. PATIENTS: Two independent cohorts of patients (n = 26 and n = 19) with histologically defined pancreatic cysts. INTERVENTION: Exploratory analysis for differentially expressed metabolites between (1) nonmucinous and mucinous cysts and (2) malignant and premalignant cysts was performed in the first cohort. With the second cohort, a validation analysis of promising identified metabolites was performed. MAIN OUTCOME MEASUREMENTS: Identification of differentially expressed metabolites between clinically relevant cyst categories and their diagnostic performance (receiver operating characteristic [ROC] curve). RESULTS: Two metabolites had diagnostic significance-glucose and kynurenine. Metabolomic abundances for both were significantly lower in mucinous cysts compared with nonmucinous cysts in both cohorts (glucose first cohort P = .002, validation P = .006; and kynurenine first cohort P = .002, validation P = .002). The ROC curve for glucose was 0.92 (95% confidence interval [CI], 0.81-1.00) and 0.88 (95% CI, 0.72-1.00) in the first and validation cohorts, respectively. The ROC for kynurenine was 0.94 (95% CI, 0.81-1.00) and 0.92 (95% CI, 0.76-1.00) in the first and validation cohorts, respectively. Neither could differentiate premalignant from malignant cysts. Glucose and kynurenine levels were significantly elevated for serous cystadenomas in both cohorts. LIMITATIONS: Small sample sizes. CONCLUSION: Metabolomic profiling identified glucose and kynurenine to have potential clinical utility for differentiating mucinous from nonmucinous pancreatic cysts. These markers also may diagnose serous cystadenomas.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Líquido Quístico/metabolismo , Cistadenocarcinoma/metabolismo , Cistoadenoma/metabolismo , Glucosa/metabolismo , Quinurenina/metabolismo , Quiste Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/metabolismo , Estudios de Cohortes , Cistadenocarcinoma/diagnóstico , Cistadenocarcinoma Mucinoso/diagnóstico , Cistadenocarcinoma Mucinoso/metabolismo , Cistoadenoma/diagnóstico , Cistoadenoma Mucinoso/diagnóstico , Cistoadenoma Mucinoso/metabolismo , Cistadenoma Seroso/diagnóstico , Cistadenoma Seroso/metabolismo , Femenino , Humanos , Masculino , Metabolómica , Persona de Mediana Edad , Quiste Pancreático/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Seudoquiste Pancreático/diagnóstico , Seudoquiste Pancreático/metabolismo , Estudios Retrospectivos , Sensibilidad y Especificidad
9.
Rapid Commun Mass Spectrom ; 27(18): 2091-2098, 2013 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-23943330

RESUMEN

RATIONALE: Metabolomic profiling is a promising methodology of identifying candidate biomarkers for disease detection and monitoring. Although lung cancer is among the leading causes of cancer-related mortality worldwide, the lung tumor metabolome has not been fully characterized. METHODS: We utilized a targeted metabolomic approach to analyze discrete groups of related metabolites. We adopted a dansyl [5-(dimethylamino)-1-naphthalene sulfonamide] derivatization with liquid chromatography/mass spectrometry (LC/MS) to analyze changes of metabolites from paired tumor and normal lung tissues. Identification of dansylated dipeptides was confirmed with synthetic standards. A systematic analysis of retention times was required to reliably identify isobaric dipeptides. We validated our findings in a separate sample cohort. RESULTS: We produced a database of the LC retention times and MS/MS spectra of 361 dansyl dipeptides. Interpretation of the spectra is presented. Using this standard data, we identified a total of 279 dipeptides in lung tumor tissue. The abundance of 90 dipeptides was selectively increased in lung tumor tissue compared to normal tissue. In a second set of validation tissues, 12 dipeptides were selectively increased. CONCLUSIONS: A systematic evaluation of certain metabolite classes in lung tumors may identify promising disease-specific metabolites. Our database of all possible dipeptides will facilitate ongoing translational applications of metabolomic profiling as it relates to lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Dipéptidos/química , Neoplasias Pulmonares/metabolismo , Metabolómica/métodos , Espectrometría de Masas en Tándem/métodos , Biomarcadores/química , Biomarcadores/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/química , Estudios de Cohortes , Dipéptidos/metabolismo , Humanos , Neoplasias Pulmonares/química
10.
Clin Transl Sci ; 16(2): 216-223, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36305236

RESUMEN

Administration of a widely used 5-hydroxytryptamine receptor (5HT3A R) antagonist (ondansetron) potently inhibited the development of experimentally induced opioid dependence and withdrawal responses in mice and humans. However, in several studies examining withdrawal symptoms in subjects with chronic opioid use disorders (OUDs), ondansetron exhibited reduced or absent efficacy. Because attenuation of opioid withdrawal symptomatology is mediated within the brain, this study examined single-dose ondansetron pharmacokinetics in the blood and brain of mice. We demonstrate that ondansetron concentrations in the brain (Cbrain ng/mg) are 1000-fold lower than the blood concentrations (Cblood ng/ml) and decrease rapidly after ondansetron administration; and that a large percentage of brain ondansetron remains in the ventricular fluid. These results indicate that the ondansetron dose, and the time window between ondansetron and opioid administration, and when withdrawal is assessed are critical considerations for clinical studies involving subjects with chronic OUD. The pharmacokinetic results and the dosing considerations discussed here can be used to improve the design of subsequent clinical trials, which will test whether a more prolonged period of ondansetron administration can provide a desperately needed therapy that can prevent the development of neonatal opioid withdrawal syndrome in babies born to mothers with chronic OUD.


Asunto(s)
Alcaloides Opiáceos , Trastornos Relacionados con Opioides , Síndrome de Abstinencia a Sustancias , Humanos , Ratones , Animales , Ondansetrón , Analgésicos Opioides , Alcaloides Opiáceos/uso terapéutico , Encéfalo , Morfina , Trastornos Relacionados con Opioides/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico
11.
J Perinatol ; 43(3): 271-276, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36030327

RESUMEN

OBJECTIVE: To determine if treatment with a 5-HT3 antagonist (ondansetron) reduces need for opioid therapy in infants at risk for neonatal opioid withdrawal syndrome (NOWS). STUDY DESIGN: A multicenter, randomized, placebo controlled, double blind clinical trial of ninety (90) infants. The intervention arms were intravenous ondansetron or placebo during labor followed by a daily dose of ondansetron or placebo in infants for five days. RESULTS: Twenty-two (49%) ondansetron-treated and 26 (63%) placebo-treated infants required pharmacologic treatment (p > 0.05). The Finnegan score was lower in the ondansetron-treated group (4.6 vs. 5.6, p = 0.02). A non-significant trend was noted for the duration of hospitalization. There was no difference in need for phenobarbital or clonidine therapy, or total dose of morphine in the first 15 days of NOWS treatment. CONCLUSIONS: Ondansetron treatment reduced the severity of NOWS symptoms; and there was an indication that it could reduce the length of stay. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov NCT01965704.


Asunto(s)
Analgésicos Opioides , Síndrome de Abstinencia Neonatal , Recién Nacido , Humanos , Analgésicos Opioides/uso terapéutico , Ondansetrón/uso terapéutico , Morfina/efectos adversos , Síndrome de Abstinencia Neonatal/tratamiento farmacológico , Fenobarbital/uso terapéutico
12.
Cancer Discov ; 13(2): 496-515, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36355448

RESUMEN

Isocitrate dehydrogenase 1 and 2 (IDH) are mutated in multiple cancers and drive production of (R)-2-hydroxyglutarate (2HG). We identified a lipid synthesis enzyme [acetyl CoA carboxylase 1 (ACC1)] as a synthetic lethal target in mutant IDH1 (mIDH1), but not mIDH2, cancers. Here, we analyzed the metabolome of primary acute myeloid leukemia (AML) blasts and identified an mIDH1-specific reduction in fatty acids. mIDH1 also induced a switch to b-oxidation indicating reprogramming of metabolism toward a reliance on fatty acids. Compared with mIDH2, mIDH1 AML displayed depletion of NADPH with defective reductive carboxylation that was not rescued by the mIDH1-specific inhibitor ivosidenib. In xenograft models, a lipid-free diet markedly slowed the growth of mIDH1 AML, but not healthy CD34+ hematopoietic stem/progenitor cells or mIDH2 AML. Genetic and pharmacologic targeting of ACC1 resulted in the growth inhibition of mIDH1 cancers not reversible by ivosidenib. Critically, the pharmacologic targeting of ACC1 improved the sensitivity of mIDH1 AML to venetoclax. SIGNIFICANCE: Oncogenic mutations in both IDH1 and IDH2 produce 2-hydroxyglutarate and are generally considered equivalent in terms of pathogenesis and targeting. Using comprehensive metabolomic analysis, we demonstrate unexpected metabolic differences in fatty acid metabolism between mutant IDH1 and IDH2 in patient samples with targetable metabolic interventions. See related commentary by Robinson and Levine, p. 266. This article is highlighted in the In This Issue feature, p. 247.


Asunto(s)
Isocitrato Deshidrogenasa , Leucemia Mieloide Aguda , Humanos , Glutaratos/metabolismo , Inhibidores Enzimáticos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación
13.
Pharmacogenet Genomics ; 22(12): 877-86, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23076370

RESUMEN

OBJECTIVE: To advance our understanding of disease biology, the characterization of the molecular target for clinically proven or new drugs is very important. Because of its simplicity and the availability of strains with individual deletions in all of its genes, chemogenomic profiling in yeast has been used to identify drug targets. As measurement of drug-induced changes in cellular metabolites can yield considerable information about the effects of a drug, we investigated whether combining chemogenomic and metabolomic profiling in yeast could improve the characterization of drug targets. BASIC METHODS: We used chemogenomic and metabolomic profiling in yeast to characterize the target for five drugs acting on two biologically important pathways. A novel computational method that uses a curated metabolic network was also developed, and it was used to identify the genes that are likely to be responsible for the metabolomic differences found. RESULTS AND CONCLUSION: The combination of metabolomic and chemogenomic profiling, along with data analyses carried out using a novel computational method, could robustly identify the enzymes targeted by five drugs. Moreover, this novel computational method has the potential to identify genes that are causative of metabolomic differences or drug targets.


Asunto(s)
Redes y Vías Metabólicas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Biología Computacional , Sistemas de Liberación de Medicamentos , Perfilación de la Expresión Génica , Metabolómica , Saccharomyces cerevisiae/efectos de los fármacos
14.
Front Microbiol ; 13: 1059289, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37063449

RESUMEN

Introduction: The routine clinical diagnosis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is largely restricted to real-time reverse transcription quantitative PCR (RT-qPCR), and tests that detect SARS-CoV-2 nucleocapsid antigen. Given the diagnostic delay and suboptimal sensitivity associated with these respective methods, alternative diagnostic strategies are needed for acute infection. Methods: We studied the use of a clinically validated liquid chromatography triple quadrupole method (LC/MS-MS) for detection of amino acids from plasma specimens. We applied machine learning models to distinguish between SARS-CoV-2-positive and negative samples and analyzed amino acid feature importance. Results: A total of 200 samples were tested, including 70 from individuals with COVID-19, and 130 from negative controls. The top performing model overall allowed discrimination between SARS-CoV-2-positive and negative control samples with an area under the receiver operating characteristic curve (AUC) of 0.96 (95%CI 0.91, 1.00), overall sensitivity of 0.99 (95%CI 0.92, 1.00), and specificity of 0.92 (95%CI 0.85, 0.95). Discussion: This approach holds potential as an alternative to existing methods for the rapid and accurate diagnosis of acute SARS-CoV-2 infection.

15.
Crit Care Med ; 39(11): 2518-25, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21765361

RESUMEN

OBJECTIVE: To investigate the effects of insulin infusion and increased parenteral amino acid intakes on whole body protein balance, glucose kinetics, and lipolysis in critically ill, insulin-resistant, septic adolescents. DESIGN: A single-center, randomized, crossover study. SETTING: A medicosurgical intensive care unit in a tertiary university hospital. PATIENTS: Nine critically ill, septic adolescents (age 15.0 ± 1.2 yrs, body mass index 20 ± 4 kg m(-2)) receiving total parenteral nutrition. INTERVENTIONS: Patients received total parenteral nutrition with standard (1.5 g · kg(-1) · day(-1)) and high (3.0 g · kg(-1) · day(-1)) amino acid intakes in a 2-day crossover setting, randomized to the order in which they received it. On both study days, we conducted a primed, constant, 7-hr stable isotope tracer infusion with [1-(13)C]leucine, [6,6-(2)H(2)]glucose, and [1,1,2,3,3-(2)H(5)]glycerol, in combination with a hyperinsulinemic euglycemic clamp during the last 3 hrs. MEASUREMENTS AND MAIN RESULTS: Insulin decreased protein synthesis at standard amino acid and high amino acid intakes (p < .01), while protein breakdown decreased with insulin at standard amino acid intake (p < .05) but not with the high amino acid intake. High amino acid intake improved protein balance (p < .05), but insulin did not have an additive effect. There was significant insulin resistance with an M value of ~3 (mg · kg(-1) · min(-1))/(mU · mL(-1)) which was 30% of reported normal values. At high amino acid intake, endogenous glucose production was not suppressed by insulin and lipolysis rates increased. CONCLUSION: The current recommended parenteral amino acid intakes are insufficient to maintain protein balance in insulin-resistant patients during tight glucose control. During sepsis, insulin decreases protein synthesis and breakdown, and while high amino acid intake improves protein balance, its beneficial effects may be offset by enhanced endogenous glucose production and lipolysis, raising concerns that insulin resistance may have been exacerbated and that gluconeogenesis may have been favored by high amino acid intakes. Dose-response studies on the effect of the level of amino acid intakes (protein) on energy metabolism are needed.


Asunto(s)
Aminoácidos/administración & dosificación , Enfermedad Crítica , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina , Insulina/uso terapéutico , Nutrición Parenteral/métodos , Sepsis/metabolismo , Adolescente , Glucemia/metabolismo , Estudios Cruzados , Metabolismo Energético/efectos de los fármacos , Femenino , Hospitales Universitarios , Humanos , Infusiones Intravenosas , Unidades de Cuidados Intensivos , Lipólisis/efectos de los fármacos , Masculino , Proteínas/metabolismo
16.
EBioMedicine ; 71: 103546, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34419924

RESUMEN

BACKGROUND: Respiratory virus infections are significant causes of morbidity and mortality, and may induce host metabolite alterations by infecting respiratory epithelial cells. We investigated the use of liquid chromatography quadrupole time-of-flight mass spectrometry (LC/Q-TOF) combined with machine learning for the diagnosis of influenza infection. METHODS: We analyzed nasopharyngeal swab samples by LC/Q-TOF to identify distinct metabolic signatures for diagnosis of acute illness. Machine learning models were performed for classification, followed by Shapley additive explanation (SHAP) analysis to analyze feature importance and for biomarker discovery. FINDINGS: A total of 236 samples were tested in the discovery phase by LC/Q-TOF, including 118 positive samples (40 influenza A 2009 H1N1, 39 influenza H3 and 39 influenza B) as well as 118 age and sex-matched negative controls with acute respiratory illness. Analysis showed an area under the receiver operating characteristic curve (AUC) of 1.00 (95% confidence interval [95% CI] 0.99, 1.00), sensitivity of 1.00 (95% CI 0.86, 1.00) and specificity of 0.96 (95% CI 0.81, 0.99). The metabolite most strongly associated with differential classification was pyroglutamic acid. Independent validation of a biomarker signature based on the top 20 differentiating ion features was performed in a prospective cohort of 96 symptomatic individuals including 48 positive samples (24 influenza A 2009 H1N1, 5 influenza H3 and 19 influenza B) and 48 negative samples. Testing performed using a clinically-applicable targeted approach, liquid chromatography triple quadrupole mass spectrometry, showed an AUC of 1.00 (95% CI 0.998, 1.00), sensitivity of 0.94 (95% CI 0.83, 0.98), and specificity of 1.00 (95% CI 0.93, 1.00). Limitations include lack of sample suitability assessment, and need to validate these findings in additional patient populations. INTERPRETATION: This metabolomic approach has potential for diagnostic applications in infectious diseases testing, including other respiratory viruses, and may eventually be adapted for point-of-care testing. FUNDING: None.


Asunto(s)
Gripe Humana/diagnóstico , Aprendizaje Automático , Metaboloma , Técnicas de Diagnóstico Molecular/métodos , Adolescente , Adulto , Niño , Preescolar , Femenino , Cromatografía de Gases y Espectrometría de Masas/métodos , Humanos , Gripe Humana/metabolismo , Gripe Humana/virología , Masculino , Metabolómica/métodos , Mucosa Nasal/metabolismo , Mucosa Nasal/virología , Orthomyxoviridae/patogenicidad , Ácido Pirrolidona Carboxílico/análisis
17.
Nat Commun ; 12(1): 6138, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686668

RESUMEN

To investigate the pathogenesis of a congenital form of hepatic fibrosis, human hepatic organoids were engineered to express the most common causative mutation for Autosomal Recessive Polycystic Kidney Disease (ARPKD). Here we show that these hepatic organoids develop the key features of ARPKD liver pathology (abnormal bile ducts and fibrosis) in only 21 days. The ARPKD mutation increases collagen abundance and thick collagen fiber production in hepatic organoids, which mirrors ARPKD liver tissue pathology. Transcriptomic and other analyses indicate that the ARPKD mutation generates cholangiocytes with increased TGFß pathway activation, which are actively involved stimulating myofibroblasts to form collagen fibers. There is also an expansion of collagen-producing myofibroblasts with markedly increased PDGFRB protein expression and an activated STAT3 signaling pathway. Moreover, the transcriptome of ARPKD organoid myofibroblasts resemble those present in commonly occurring forms of liver fibrosis. PDGFRB pathway involvement was confirmed by the anti-fibrotic effect observed when ARPKD organoids were treated with PDGFRB inhibitors. Besides providing insight into the pathogenesis of congenital (and possibly acquired) forms of liver fibrosis, ARPKD organoids could also be used to test the anti-fibrotic efficacy of potential anti-fibrotic therapies.


Asunto(s)
Cirrosis Hepática/patología , Modelos Biológicos , Organoides/patología , Enfermedades de los Conductos Biliares/genética , Enfermedades de los Conductos Biliares/metabolismo , Enfermedades de los Conductos Biliares/patología , Colágeno/metabolismo , Células Epiteliales/patología , Humanos , Células Madre Pluripotentes Inducidas/citología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Mutación , Miofibroblastos/metabolismo , Miofibroblastos/patología , Organoides/efectos de los fármacos , Organoides/metabolismo , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/patología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
18.
Am J Physiol Endocrinol Metab ; 297(5): E1046-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19724018

RESUMEN

To determine the rates of methionine splanchnic uptake and utilization in critically ill pediatric patients we used two kinetic models: the plasma methionine enrichment and the "intracellular" homocysteine enrichment. Twenty four patients, eight infants, eight children, and eight adolescents, were studied. They received simultaneous, primed, constant, intravenous infusions of l-[(2)H(3)]methylmethionine and enteral l-[1-(13)C]methionine. The ratio of [(13)C]homocysteine to [(13)C]methionine enrichment was 1.0 ± 0.15, 0.80 ± 0.20, and 0.66 ± 0.10, respectively, for the infants, children, and adolescents, and it was different between the infants and adolescents (P < 0.01). Methionine splanchnic uptake was 63, 45, and 36%, respectively, in the infants, children, and adolescents, and it was higher (P < 0.01) in the infants compared with the adolescents. The infants utilized 73% of methionine flux for nonoxidative disposal, while 27% was used for transulfuration (P < 0.001). Conversely, in the adolescents, 40% was utilized for nonoxidative disposal, while 60% was used for transulfuration. There is ontogeny on the rates of methionine splanchnic uptake and on the fate of methionine utilization in critically ill children, with greater methionine utilization for synthesis of proteins and methionine-derived compounds (P < 0.01) and decreased transulfuration rates in the infants (P < 0.01), while the opposite was observed in the adolescents. The plasma model underestimated methionine kinetics in children and adolescents but not in the infants, suggesting lesser dilution and greater compartmentation of methionine metabolism in the infant population. All patients were in negative methionine balance, indicating that the current enteral nutritional support is inadequate in these patients.


Asunto(s)
Abdomen/fisiología , Enfermedad Crítica , Metionina/metabolismo , Adolescente , Envejecimiento/metabolismo , Algoritmos , Dióxido de Carbono/metabolismo , Niño , Preescolar , Dieta , Nutrición Enteral , Femenino , Homocisteína/metabolismo , Humanos , Lactante , Marcaje Isotópico , Cinética , Masculino , Metionina/farmacocinética , Oxidación-Reducción
19.
Clin Sci (Lond) ; 117(1): 23-30, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19105791

RESUMEN

Arginine has vasodilatory effects, via its conversion by NO synthase into NO, and immunomodulatory actions which play important roles in sepsis. Protein breakdown affects arginine availability and the release of asymmetric dimethylarginine, an inhibitor of NO synthase, may therefore affect NO synthesis in patients with sepsis. The objective of the present study was to investigate whole-body in vivo arginine and citrulline metabolism and NO synthesis rates, and their relationship to protein breakdown in patients with sepsis or septic shock and in healthy volunteers. Endogenous leucine flux, an index of whole-body protein breakdown rate, was measured in 13 critically ill patients with sepsis or septic shock and seven healthy controls using an intravenous infusion of [1-13C]leucine. Arginine flux, citrulline flux and the rate of conversion of arginine into citrulline (an index of NO synthesis) were measured with intravenous infusions of [15N2]guanidino-arginine and [5,5-2H2]citrulline. Plasma concentrations of nitrite plus nitrate, arginine, citrulline and asymmetric dimethylarginine were measured. Compared with controls, patients had a higher leucine flux and higher NO metabolites, but arginine flux, plasma asymmetric dimethylarginine concentration and the rate of NO synthesis were not different. Citrulline flux and plasma arginine and citrulline were lower in patients than in controls. Arginine production was positively correlated with the protein breakdown rate. Whole-body arginine production and NO synthesis were similar in patients with sepsis and septic shock and healthy controls. Despite increased proteolysis in sepsis, there is a decreased arginine plasma concentration, suggesting inadequate de novo synthesis secondary to decreased citrulline production.


Asunto(s)
Arginina/metabolismo , Citrulina/metabolismo , Óxido Nítrico/biosíntesis , Choque Séptico/metabolismo , Arginina/análogos & derivados , Estudios de Casos y Controles , Femenino , Homeostasis , Humanos , Leucina/metabolismo , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA